Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Mol Nutr Food Res ; 57(2): 277-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23197441

ABSTRACT

SCOPE: Epidemiological and experimental evidence indicates that maternal nutrition status contributes to long-term changes in the metabolic phenotype of the offspring, a process known as fetal programming. METHODS AND RESULTS: We have used a swine model (Sus scrofa) to analyze consequences of a maternal low protein diet (about 50% of control) during pregnancy on hepatic lipid metabolism and genome-wide hepatic gene expression profile of juvenile female offspring (mean age 85 days). We found 318 S. scrofa genes to be differentially expressed in the liver at age 85 days. In the low protein offspring group key genes of fatty acid de novo synthesis were downregulated whereas several genes of lipolysis and phospholipid biosynthesis were upregulated. qRT-PCR analysis of selected genes verified microarray data and revealed linear correlations between gene expression levels and slaughter weight. Hepatic cholesterol 7α hydroxylase protein expression tended to be lower in the low protein group. Total lipid and triglyceride content and fatty acid composition of total lipids were not different between groups. CONCLUSION: A maternal low protein diet during pregnancy induces a distinct hepatic gene expression signature in juvenile female pigs which was not translated into phenotypical changes of liver lipid metabolism.


Subject(s)
Diet, Protein-Restricted , Liver/metabolism , Maternal Nutritional Physiological Phenomena , Transcriptome , Animals , Blotting, Western , Body Weight , Chromatography, Gas , Chromatography, Thin Layer , Computational Biology , Dietary Proteins/administration & dosage , Female , Fetal Development , Gene Expression Regulation , Lipid Metabolism/drug effects , Malnutrition/physiopathology , Phenotype , Pregnancy , Real-Time Polymerase Chain Reaction , Swine , Triglycerides/blood
2.
BMC Res Notes ; 5: 540, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23021568

ABSTRACT

BACKGROUND: Coenzyme Q10 is an essential cofactor in the respiratory chain and serves in its reduced form, ubiquinol, as a potent antioxidant. Studies in vitro and in vivo provide evidence that ubiquinol reduces inflammatory processes via gene expression. Here we investigate the putative link between expression and DNA methylation of ubiquinol sensitive genes in monocytes obtained from human volunteers supplemented with 150 mg/ day ubiquinol for 14 days. FINDINGS: Ubiquinol decreases the expression of the pro-inflammatory chemokine (C-X-C motif) ligand 2 gene (CXCL2) more than 10-fold. Bisulfite-/ MALDI-TOF-based analysis of regulatory regions of the CXCL2 gene identified six adjacent CpG islands which showed a 3.4-fold decrease of methylation status after ubiquinol supplementation. This effect seems to be rather gene specific, because ubiquinol reduced the expression of two other pro-inflammatory genes (PMAIP1, MMD) without changing the methylation pattern of the respective gene. CONCLUSION: In conclusion, ubiquinol decreases monocytic expression and DNA methylation of the pro-inflammatory CXCL2 gene in humans. Current Controlled Trials ISRCTN26780329.


Subject(s)
Chemokine CXCL2/metabolism , DNA Methylation/drug effects , Dietary Supplements , Inflammation Mediators/metabolism , Monocytes/drug effects , Ubiquinone/analogs & derivatives , Adult , Chemokine CXCL2/genetics , CpG Islands/drug effects , Down-Regulation , Humans , Male , Middle Aged , Monocytes/metabolism , RNA, Messenger/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Ubiquinone/pharmacology , Young Adult
3.
J Clin Biochem Nutr ; 50(2): 119-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22448092

ABSTRACT

Coenzyme Q(10) is an essential cofactor in the respiratory chain and serves as a potent antioxidant in biological membranes. Recent studies in vitro and in vivo provide evidence that Coenzyme Q(10) is involved in inflammatory processes and lipid metabolism via gene expression. To study these effects at the epigenomic level, C57BL6J mice were supplemented for one week with reduced Coenzyme Q(10) (ubiquinol). Afterwards, gene expression signatures and DNA promoter methylation patterns of selected genes were analysed. Genome-wide transcript profiling in the liver identified 1112 up-regulated and 571 down-regulated transcripts as differentially regulated between ubiquinol-treated and control animals. Text mining and GeneOntology analysis revealed that the "top 20" ubiquinol-regulated genes play a role in lipid metabolism and are functionally connected by the PPARα signalling pathway. With regard to the ubiquinol-induced changes in gene expression of about +3.14-fold (p≤0.05), +2.18-fold (p≤0.01), and -2.13-fold (p≤0.05) for ABCA1, ACYP1, and ACSL1 genes, respectively, hepatic DNA methylation analysis of 282 (sense orientation) and 271 (antisense) CpG units in the respective promoter islands revealed no significant effect of ubiquinol. In conclusion, ubiquinol affects the expression of genes involved in PPARα signalling and lipid metabolism without changing the promoter DNA methylation status in the liver of mice.

4.
Mutat Res ; 733(1-2): 61-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-21964355

ABSTRACT

Coenzyme Q(10) (CoQ(10)) is an essential component for electron transport in the mitochondrial respiratory chain and serves as cofactor in several biological processes. The reduced form of CoQ(10) (ubiquinol, Q(10)H(2)) is an effective antioxidant in biological membranes. During the last years, particular interest has been grown on molecular effects of CoQ(10) supplementation on mechanisms related to DNA damage prevention. This review describes recent advances in our understanding about the impact of CoQ(10) on genomic stability in cells, animals and humans. With regard to several in vitro and in vivo studies, CoQ(10) provides protective effects on several markers of oxidative DNA damage and genomic stability. In comparison to the number of studies reporting preventive effects of CoQ(10) on oxidative stress biomarkers, CoQ(10) intervention studies in humans with a direct focus on markers of DNA damage are limited. Thus, more well-designed studies in healthy and disease populations with long-term follow up results are needed to substantiate the reported beneficial effects of CoQ(10) on prevention of DNA damage.


Subject(s)
DNA Damage , Ubiquinone/analogs & derivatives , Animals , Cells, Cultured , Dietary Supplements , Genomic Instability , Humans , Lipid Peroxidation , Micronutrients/genetics , Oxidative Stress , Ubiquinone/metabolism , Ubiquinone/physiology
5.
BMC Res Notes ; 4: 245, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21774831

ABSTRACT

BACKGROUND: Coenzyme Q10 (CoQ10) is essential for mitochondrial energy production and serves as an antioxidants in extra mitochondrial membranes. The genetics of primary CoQ10 deficiency has been described in several studies, whereas the influence of common genetic variants on CoQ10 status is largely unknown. Here we tested for non-synonymous single-nucleotidepolymorphisms (SNP) in genes involved in the biosynthesis (CoQ3G272S , CoQ6M406V, CoQ7M103T), reduction (NQO1P187S, NQO2L47F) and metabolism (apoE3/4) of CoQ10 and their association with CoQ10 status. For this purpose, CoQ10 serum levels of 54 healthy male volunteers were determined before (T0) and after a 14 days supplementation (T14) with 150 mg/d of the reduced form of CoQ10. FINDINGS: At T0, the CoQ10 level of heterozygous NQO1P187S carriers were significantly lower than homozygous S/S carriers (0.93 ± 0.25 µM versus 1.34 ± 0.42 µM, p = 0.044). For this polymorphism a structure homology-based method (PolyPhen) revealed a possibly damaging effect on NQO1 protein activity. Furthermore, CoQ10 plasma levels were significantly increased in apoE4/E4 genotype after supplementation in comparison to apoE2/E3 genotype (5.93 ± 0.151 µM versus 4.38 ± 0.792 µM, p = 0.034). Likewise heterozygous CoQ3G272S carriers had higher CoQ10 plasma levels at T14 compared to G/G carriers but this difference did not reach significance (5.30 ± 0.96 µM versus 4.42 ± 1.67 µM, p = 0.082). CONCLUSIONS: In conclusion, our pilot study provides evidence that NQO1P187S and apoE polymorphisms influence CoQ10 status in humans.

6.
BMC Res Notes ; 4: 141, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21569287

ABSTRACT

BACKGROUND: Human paraoxonase 1 (PON1) is an HDL-associated enzyme with anti-oxidant/anti-inflammatory properties that has been suggested to play an important protective role against coronary heart diseases and underlying atherogenesis. The common PON1 Q192R polymorphism (rs662, A>G), a glutamine to arginine substitution at amino acid residue 192, has been analyzed in numerous association studies as a genetic marker for coronary heart diseases, however, with controversial results. FINDINGS: To get a better understanding about the pathophysiological function of PON1, we analyzed the relationships between the Q192R polymorphism, serum paraoxonase activity and serum biomarkers important for atherogenesis. Genotyping a cohort of 49 healthy German males for the Q192R polymorphism revealed an allele distribution of 0.74 and 0.26 for the Q and R allele, respectively, typical for Caucasian populations. Presence of the R192 allele was found to be associated with a significantly increased paraoxonase enzyme activity of 187.8 ± 11.4 U/l in comparison to the QQ192 genotype with 60.5 ± 4.9 U/l. No significant differences among the genotypes were found for blood pressure, asymmetric dimethylarginine, LDL, HDL, triglycerides, and cholesterol. As expected, MIP-2 alpha a cytokine rather not related to atherosclerosis is not affected by the PON1 polymorphism. In contrast to that, the pro-inflammatory cytokine TNF-alpha is enhanced in R192 carriers (163.8 ± 24.7 pg/ml vs 94.7 ± 3.2 pg/ml in QQ192 carriers). CONCLUSIONS: Our findings support the hypothesis that the common PON1 R192 allele may be a genetic risk factor for atherogenesis by inducing chronic low-grade inflammation.

7.
J Med Food ; 14(4): 391-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21370964

ABSTRACT

Monocytes are key players in inflammatory processes that are triggered by lipopolysaccharide (LPS), the major outer membrane component of Gram-negative bacteria. The present study in human monocytic THP-1 cells was designed in order to identify LPS-inducible genes that are down-regulated by the reduced form of coenzyme Q(10) (ubiquinol, Q(10)H(2)). For this purpose, THP-1 cells were incubated with 10 µM Q(10)H(2) for 24 hours. Subsequently, cells were stimulated for 4 hours with 1 µg/mL LPS, and the resulting gene expression levels were determined using microarrays. Fourteen LPS-inducible genes were identified to be significantly (P ≤ .05) down-regulated by Q(10)H(2) pretreatment between a factor of 1.32 and 1.65. The strongest effect of Q(10)H(2) incubation was found for the nuclear receptor coactivator 2 gene (NCOA2). Gene ontology terms revealed for the Q(10)H(2)-sensitive genes an involvement in, e.g., signal transduction processes (centaurin, delta 1; NCOA2; pleckstrin and Sec7 domain containing 3; protein phosphatase 2, regulatory subunit B [B56], γ isoform), transcriptional regulation (NCOA2; POU domain, class 2, transcription factor 1; ETS variant gene 3), and cell proliferation pathways (hypothetical protein FLJ36090, epidermal growth factor receptor pathway substrate 15). In conclusion, we provide evidence in THP-1 cells that Q(10)H(2) modulates LPS-induced gene expression.


Subject(s)
Down-Regulation , Lipopolysaccharides/metabolism , Nuclear Receptor Coactivator 2/genetics , Ubiquinone/analogs & derivatives , Cell Line , Cell Proliferation , Gene Expression , Gene Expression Profiling , Humans , Monocytes/drug effects , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , Signal Transduction , Ubiquinone/pharmacology
8.
IUBMB Life ; 63(1): 42-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21280176

ABSTRACT

Studies in vitro and in mice indicate a role for Coenzyme Q(10) (CoQ(10) ) in gene expression. To determine this function in relationship to physiological readouts, a 2-week supplementation study with the reduced form of CoQ(10) (ubiquinol, Q(10) H(2) , 150 mg/d) was performed in 53 healthy males. Mean CoQ(10) plasma levels increased 4.8-fold after supplementation. Transcriptomic and bioinformatic approaches identified a gene-gene interaction network in CD14-positive monocytes, which functions in inflammation, cell differentiation, and peroxisome proliferator-activated receptor-signaling. These Q(10) H(2) -induced gene expression signatures were also described previously in liver tissues of SAMP1 mice. Biochemical and NMR-based analyses showed a reduction of low density lipoprotein (LDL) cholesterol plasma levels after Q(10) H(2) supplementation. This effect was especially pronounced in atherogenic small dense LDL particles (19-21 nm, 1.045 g/L). In agreement with gene expression signatures, Q(10) H(2) reduces the number of erythrocytes but increases the concentration of reticulocytes. In conclusion, Q(10) H(2) induces characteristic gene expression patterns, which are translated into reduced LDL cholesterol levels and altered parameters of erythropoiesis in humans.


Subject(s)
Cholesterol, LDL/blood , Erythropoiesis/drug effects , Gene Expression/drug effects , Ubiquinone/analogs & derivatives , Adult , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Humans , Magnetic Resonance Spectroscopy , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Ubiquinone/pharmacology
9.
BMC Complement Altern Med ; 11: 1, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-21199573

ABSTRACT

BACKGROUND: Both resveratrol and vitamin C (ascorbic acid) are frequently used in complementary and alternative medicine. However, little is known about the underlying mechanisms for potential health benefits of resveratrol and its interactions with ascorbic acid. METHODS: The antioxidant enzymes heme oxygenase-1 and paraoxonase-1 were analysed for their mRNA and protein levels in HUH7 liver cells treated with 10 and 25 µmol/l resveratrol in the absence and presence of 100 and 1000 µmol/l ascorbic acid. Additionally the transactivation of the transcription factor Nrf2 and paraoxonase-1 were determined by reporter gene assays. RESULTS: Here, we demonstrate that resveratrol induces the antioxidant enzymes heme oxygenase-1 and paraoxonase-1 in cultured hepatocytes. Heme oxygenase-1 induction by resveratrol was accompanied by an increase in Nrf2 transactivation. Resveratrol mediated Nrf2 transactivation as well as heme oxygenase-1 induction were partly antagonized by 1000 µmol/l ascorbic acid. CONCLUSIONS: Unlike heme oxygenase-1 (which is highly regulated by Nrf2) paraoxonase-1 (which exhibits fewer ARE/Nrf2 binding sites in its promoter) induction by resveratrol was not counteracted by ascorbic acid. Addition of resveratrol to the cell culture medium produced relatively low levels of hydrogen peroxide which may be a positive hormetic redox-signal for Nrf2 dependent gene expression thereby driving heme oxygenase-1 induction. However, high concentrations of ascorbic acid manifold increased hydrogen peroxide production in the cell culture medium which may be a stress signal thereby disrupting the Nrf2 signalling pathway.


Subject(s)
Antioxidants/metabolism , Aryldialkylphosphatase/metabolism , Ascorbic Acid/pharmacology , Heme Oxygenase-1/metabolism , Hepatocytes/drug effects , Plant Extracts/pharmacology , Stilbenes/pharmacology , Antioxidants/pharmacology , Aryldialkylphosphatase/genetics , Ascorbic Acid/administration & dosage , Carcinoma, Hepatocellular , Cell Line, Tumor , Drug Interactions , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/genetics , Hepatocytes/enzymology , Humans , Hydrogen Peroxide/metabolism , NF-E2-Related Factor 2/metabolism , Plant Extracts/administration & dosage , RNA, Messenger/metabolism , Resveratrol , Transcriptional Activation/drug effects
10.
IUBMB Life ; 62(11): 812-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21086475

ABSTRACT

Studies in humans and mice indicate a role for coenzyme Q(10) (CoQ(10)) in gene expression. To analyze this function in relation to metabolism, SAMP1 mice were supplemented with the reduced (ubiquinol) or oxidized (ubiquinone) form of CoQ(10) (500 mg/kg BW/d) for 14 months. Microarray analyses in liver tissues of SAMP1 mice identified 946 genes as differentially expressed between ubiquinol-treated and control animals (≥1.5-fold, P < 0.05). Text mining analyses revealed for a part of the ubiquinol-regulated genes, a functional connection in PPARα and LXR/RXR signalling pathways. Because these pathways are involved in cholesterol homeostasis, relevant metabolites were determined by gas chromatography/mass spectrometry (GC/MS). We found a significant increase of desmosterol (2.0-fold, P < 0.001) in the liver of ubiquinol-supplemented SAMP1 mice when related to control animals. In agreement, cholesterol concentrations were also distinctly increased (1.3-fold, P = 0.057). The Q(10)H(2)-induced PPARα and LXR/RXR gene expression signatures and effects on cholesterol metabolism were not apparent for the oxidized form of CoQ(10). In conclusion, the reduced form of CoQ(10) mediates distinct effects on cholesterol metabolism at the transcriptional and metabolite level in SAMP1 mice.


Subject(s)
Cholesterol/metabolism , Liver/metabolism , Ubiquinone/analogs & derivatives , Animals , Desmosterol/metabolism , Female , Gas Chromatography-Mass Spectrometry , Gene Expression Profiling , Mice , Oxidation-Reduction , PPAR alpha/metabolism , Retinoid X Receptors/metabolism , Ubiquinone/pharmacology
11.
Biofactors ; 36(3): 222-8, 2010.
Article in English | MEDLINE | ID: mdl-20533395

ABSTRACT

Coenzyme Q(10) (CoQ(10)) is an obligatory element in the respiratory chain and functions as a potent antioxidant of lipid membranes. More recently, anti-inflammatory effects as well as an impact of CoQ(10) on gene expression have been observed. To reveal putative effects of Q(10) on LPS-induced gene expression, whole genome expression analysis was performed in the monocytic cell line THP-1. Thousand one hundred twenty-nine and 710 probe sets have been identified to be significantly (P

Subject(s)
Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Ubiquinone/pharmacology , Cell Line , Down-Regulation , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Micronutrients/pharmacology , Oligonucleotide Array Sequence Analysis
12.
Mol Nutr Food Res ; 54(6): 805-15, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19960455

ABSTRACT

Our present study reveals significant decelerating effects on senescence processes in middle-aged SAMP1 mice supplemented for 6 or 14 months with the reduced form (Q(10)H(2), 500 mg/kg BW/day) of coenzyme Q(10) (CoQ(10)). To unravel molecular mechanisms of these CoQ(10) effects, a genome-wide transcript profiling in liver, heart, brain and kidney of SAMP1 mice supplemented with the reduced (Q(10)H(2)) or oxidized form of CoQ(10) (Q(10)) was performed. Liver seems to be the main target tissue of CoQ(10) intervention, followed by kidney, heart and brain. Stringent evaluation of the resulting data revealed that Q(10)H(2) has a stronger impact on gene expression than Q(10), primarily due to differences in the bioavailability. Indeed, Q(10)H(2) supplementation was more effective than Q(10) to increase levels of CoQ(10) in the liver of SAMP1 mice. To identify functional and regulatory connections of the "top 50" (p<0.05) Q(10)H(2)-sensitive transcripts in liver, text mining analysis was used. Hereby, we identified Q(10)H(2)-sensitive genes which are regulated by peroxisome proliferator-activated receptor-alpha and are primarily involved in cholesterol synthesis (e.g. HMGCS1, HMGCL and HMGCR), fat assimilation (FABP5), lipoprotein metabolism (PLTP) and inflammation (STAT-1). These data may explain, at least in part, the decelerating effects on degenerative processes observed in Q(10)H(2)-supplemented SAMP1 mice.


Subject(s)
Aging/drug effects , PPAR alpha/genetics , Ubiquinone/analogs & derivatives , Animals , Dietary Supplements , Eating/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Liver/metabolism , Mice , Oxidation-Reduction , Phenotype , Ubiquinone/metabolism , Ubiquinone/pharmacology
13.
Mediators Inflamm ; 2009: 415437, 2009.
Article in English | MEDLINE | ID: mdl-19390647

ABSTRACT

MicroRNAs (miRs) are involved in key biological processes via suppression of gene expression at posttranscriptional levels. According to their superior functions, subtle modulation of miR expression by certain compounds or nutrients is desirable under particular conditions. Bacterial lipopolysaccharide (LPS) induces a reactive oxygen species-/NF-kappaB-dependent pathway which increases the expression of the anti-inflammatory miR-146a. We hypothesized that this induction could be modulated by the antioxidant ubiquinol-10. Preincubation of human monocytic THP-1 cells with ubiquinol-10 reduced the LPS-induced expression level of miR-146a to 78.9 +/- 13.22%. In liver samples of mice injected with LPS, supplementation with ubiquinol-10 leads to a reduction of LPS-induced miR-146a expression to 78.12 +/- 21.25%. From these consistent in vitro and in vivo data, we conclude that ubiquinol-10 may fine-tune the inflammatory response via moderate reduction of miR-146a expression.


Subject(s)
Antioxidants/pharmacology , MicroRNAs/drug effects , MicroRNAs/metabolism , Ubiquinone/analogs & derivatives , Animals , Cell Line , Down-Regulation , Humans , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Ubiquinone/pharmacology , Up-Regulation
14.
J Clin Biochem Nutr ; 44(1): 62-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19177190

ABSTRACT

Ubiquinol-10 (QH(2)), the reduced form of Coenzyme Q(10) (CoQ(10)) serves as a potent antioxidant of lipid membranes. Because many antioxidants reveal potent anti-inflammatory effects, the influence of QH(2) on lipopolysaccharide (LPS)-induced pro-inflammatory cytokines and chemokines were determined in the human monocytic cell line THP-1. Stimulation of cells with LPS resulted in a distinct release of Tumour necrosis factor-alpha (TNF-alpha), Macrophage inflammatory protein-1 alpha (MIP-1alpha), Regulated upon activation, normal T cell expressed and secreted (RANTES) and Monocyte chemotattractant protein-1 (MCP-1). The LPS-induced responses were significantly decreased by pre-incubation of cells with QH(2) to 60.27 +/- 9.3% (p = 0.0009), 48.13 +/- 6.93% (p = 0.0007) and 74.36 +/- 7.25% (p = 0.008) for TNF-alpha, MIP-1alpha and RANTES, respectively. In conclusion, our results indicate anti-inflammatory effects of the reduced form of CoQ(10) on various proinflammatory cytokines and chemokines in vitro.

15.
Biofactors ; 32(1-4): 179-83, 2008.
Article in English | MEDLINE | ID: mdl-19096114

ABSTRACT

Clinical studies demonstrated the efficacy of Coenzyme Q10 (CoQ10) as an adjuvant therapeutic in cardiovascular diseases, mitochondrial myopathies and neurodegenerative diseases. More recently, expression profiling revealed that Coenzyme Q10 (CoQ10) influences the expression of several hundred genes. To unravel the functional connections of these genes, we performed a text mining approach using the Genomatix BiblioSphere. We identified signalling pathways of G-protein coupled receptors, JAK/STAT, and Integrin which contain a number of CoQ10 sensitive genes. Further analysis suggested that IL5, thrombin, vitronectin, vitronectin receptor, and C-reactive protein are regulated by CoQ10 via the transcription factor NFkappaB1. To test this hypothesis, we studied the effect of CoQ10 on the NFkappaB1-dependent pro-inflammatory cytokine TNF-alpha. As a model, we utilized the murine macrophage cell lines RAW264.7 transfected with human apolipoprotein E3 (apoE3, control) or pro-inflammatory apoE4. In the presence of 2.5 microM or 75 microM CoQ10 the LPS-induced TNF-alpha response was significantly reduced to 73.3 +/- 2.8% and 74.7 +/- 8.9% in apoE3 or apoE4 cells, respectively. Therefore, the in silico analysis as well as the cell culture experiments suggested that CoQ10 exerts anti-inflammatory properties via NFkappaB1-dependent gene expression.


Subject(s)
Gene Expression/drug effects , Inflammation/physiopathology , Ubiquinone/analogs & derivatives , Animals , Cell Line , Computer Simulation , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/physiology , Mice , Models, Biological , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Ubiquinone/physiology
16.
J Athl Train ; 43(5): 489-504, 2008.
Article in English | MEDLINE | ID: mdl-18833312

ABSTRACT

CONTEXT: Elite distance runners (EDR) must optimize their nutrition to maintain their demanding training schedules. OBJECTIVE: To develop a nutrition concept for EDR based on energy and macronutrient expenditures. DESIGN: This theoretical study provides calculations for macronutrient and energy expenditures of EDR. Anthropometric and metabolic characteristics of EDR were assumed based on average real EDR. SETTING: University of Kiel. PATIENTS OR OTHER PARTICIPANTS: Three prototypic types of male EDR described in the literature as type I (TI; body mass = 72 kg, respiratory quotient = 0.9 at rest, fast-twitch muscle fibers = 60% to 70%), type II (TII; body mass = 67 kg, respiratory quotient = 0.82 at rest, fast-twitch muscle fibers = 50%), and type III (TIII; body mass = 60 kg, respiratory quotient = 0.75 at rest, fast-twitch muscle fibers = 30% to 40%). MAIN OUTCOME MEASURE(S): We calculated the macronutrient and energy expenditures of the 3 types of EDR according to body mass, respiratory quotient, and percentage of fast-twitch muscle fibers. RESULTS: We found that the average energy expenditure was 3750 kcal . d(-1) for TI runners, 3463 kcal . d(-1) for TII runners, and 3079 kcal . d(-1) for TIII runners. The carbohydrate (CHO) expenditure reached an average value of 10.0 g . kg(-1) . d(-1) for TI runners, 8.0 g . kg(-1) . d(-1) for TII runners, and 4.7 g . kg(-1) . d(-1) for TIII runners. When the EDR accomplished running sessions at a pace >or=100% of maximum oxygen consumption, all types of runners had a CHO demand of about 10 g . kg(-1) . d(-1). The TI and TII runners need a CHO intake of 8 to 10 g . kg(-1) . d(-1). For the TIII runners, a CHO intake >6 g . kg(-1) . d(-1) is necessary during anaerobic training sessions. CONCLUSIONS: Nutrition concepts must be differentiated for EDR according to metabolic and anthropometric characteristics of the runners and their special training emphases.


Subject(s)
Energy Intake/physiology , Energy Metabolism/physiology , Nutritional Requirements , Oxygen Consumption/physiology , Physical Endurance/physiology , Running/physiology , Adolescent , Adult , Anthropometry , Body Mass Index , Diet , Dietary Carbohydrates/metabolism , Dietary Fats/metabolism , Dietary Proteins/metabolism , Health Services Accessibility , Humans , Male , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/physiology , Young Adult
17.
FEBS Lett ; 582(23-24): 3542-6, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18817776

ABSTRACT

Dietary vitamin E (VE) is known to regulate gene expression by altering mRNA concentrations. Recently, microRNA (miRNA) have been discovered as a means of posttranscriptional gene regulation. Since the effect of VE on miRNA regulation is unknown, we fed rats for 6 months diets deficient or sufficient in VE and determined hepatic concentrations of miRNA involved in processes previously associated with VE (lipid metabolism, miRNA-122a; cancer and inflammation, miRNA-125b). VE-deficiency resulted in reduced concentrations of miRNA-122a and miRNA-125b. The findings of the present study demonstrate that differences in dietary VE may affect hepatic miRNA concentrations in vivo.


Subject(s)
Liver/metabolism , MicroRNAs/metabolism , Vitamin E/metabolism , Animals , Base Sequence , Cell Transformation, Neoplastic/genetics , Diet , Gene Expression Regulation/drug effects , Inflammation/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Male , Rats , Rats, Inbred F344 , Vitamin E/administration & dosage , Vitamin E/pharmacology
18.
IUBMB Life ; 59(10): 628-633, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17852568

ABSTRACT

Coenzyme Q10 (CoQ10, ubiquinone) is an essential cofactor in the electron transport chain, serves as a potent antioxidant in mitochondria and lipid membranes, and is often used as a dietary supplement for a number of diseases including cardiovascular diseases. Recently, we obtained evidence that CoQ10 (Kaneka Q10) affects the expression of hundreds of human genes. To decipher the functional and regulatory connections of these genes, a literature search combined with transcription factor binding site analysis was performed using Genomatix BiblioSphere and MatInspector. This in-silico analysis revealed 17 CoQ10-inducible genes which are functionally connected by signalling pathways of G-protein coupled receptors, JAK/STAT, integrin, and beta-arrestin. Promoter analysis of these CoQ10-inducible genes showed one group of NF B-regulated genes, namely IL5, thrombin, vitronectin receptor and C-reactive protein (CRP). Furthermore, a common promoter framework containing binding sites of the transcription factor families EVI1, HOXF, HOXC, and CLOX was identified in the promoters of IL5, CRP, and vitronectin receptor. The identified CoQ10-inducible genes and pathways play an important role in inflammatory response. Since these effects are based on an in-vitro study, the effect of CoQ10 on vascular health in vivo needs to be addressed in further animal and/or human intervention studies.


Subject(s)
Computational Biology , Gene Expression Regulation/physiology , Signal Transduction/genetics , Ubiquinone/analogs & derivatives , Caco-2 Cells , Coenzymes/physiology , Computational Biology/methods , Humans , Inflammation/genetics , Inflammation/metabolism , Software , Ubiquinone/physiology
19.
Biofactors ; 31(1): 35-41, 2007.
Article in English | MEDLINE | ID: mdl-18806307

ABSTRACT

Studies in humans and cell culture as well as bioinformatics suggested that Coenzyme Q(10) (CoQ10) functions as an anti-inflammatory molecule. Here we studied the influence of CoQ10 (Kaneka Q10) on secretion of the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) by using the human and murine monocytic cell lines THP-1 and RAW264.7 expressing human apolipoprotein E3 (apoE3) or pro-inflammatory apoE4. Incubation of cells with physiological (0.1-10 microM) and supra-physiological (> 10 to < 100 microM) concentrations of CoQ10 led to an intracellular accumulation of its reduced form without any cytotoxic effects. Stimulation of cell models with lipopolysaccharide (LPS) resulted in a substantially release of TNF-alpha. When THP-1 cells were pre-incubated with 10 microM CoQ10, the LPS-induced TNF-alpha release was significantly decreased to 72 +/- 32%. This effect is similar to those obtained by 10 microM N-Acetyl-Cysteine, a well known reference antioxidant. In RAW264.7-apoE3 and -apoE4 cells, significant reductions of LPS-induced TNF-alpha secretion to 73.3 +/- 2.8% and 74.7 +/- 8.9% were found with 2.5 microM and 75 microM CoQ10, respectively. In conclusion, CoQ10 has moderate anti-inflammatory effects in two monocytic cell lines which could be mediated by its antioxidant activity.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Monocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquinone/analogs & derivatives , Animals , Apolipoprotein E3/physiology , Apolipoprotein E4/physiology , Cell Line , Cell Survival/drug effects , Humans , Mice , Monocytes/drug effects , Ubiquinone/pharmacology
20.
Biofactors ; 31(3-4): 211-7, 2007.
Article in English | MEDLINE | ID: mdl-18997284

ABSTRACT

Coenzyme Q_{10} (CoQ_{10}) is an obligatory element in the mitochondrial electron transport system and functions as a potent antioxidant of lipid membranes. In-vivo and in-vitro studies indicate an involvement of CoQ_{10} in inflammatory pathways. Here we studied in the human monocytic cell-line THP-1 the influence of CoQ_{10} on LPS-induced secretion of the pro-inflammatory chemokines Macrophage inflammatory protein-1 alpha (MIP-1alpha), Regulated upon activation, normal T cell expressed and secreted (RANTES) and Monocyte chemoattractant protein-1 (MCP-1). In comparison to unstimulated cells, LPS leads to 22-, 3- and 4.5-fold higher levels of MIP-1alpha, RANTES and MCP-1 in the cell culture medium, respectively. Pre-incubation of cells with 10 microM CoQ_{10} resulted in a significant decrease of LPS-induced MIP-1alpha and RANTES secretion to 55.04% (p = 0.02) and 76.84% (p = 0.04), respectively. In conclusion, CoQ_{10} reduces the LPS-induced secretion levels of the pro-inflammatory chemokines MIP-1alpha and RANTES in the human monocytic cell line THP-1. These data suggest that CoQ_{10} possesses anti-inflammatory properties.


Subject(s)
Chemokines/metabolism , Monocytes/drug effects , Ubiquinone/analogs & derivatives , Cell Line , Cell Survival/drug effects , Chemokine CCL2/metabolism , Dose-Response Relationship, Drug , Humans , Lipopolysaccharides/pharmacology , Monocytes/cytology , Monocytes/metabolism , Ubiquinone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...